<<

View metadata, citation and similar papers at core.ac.uk brought to you by CORE

provided by Springer - Publisher Connector Benemei et al. The Journal of Headache and Pain 2013, 14:71 http://www.thejournalofheadacheandpain.com/content/14/1/71

REVIEW ARTICLE Open Access TRPA1 and other TRP channels in migraine Silvia Benemei*, Francesco De Cesaris, Camilla Fusi, Eleonora Rossi, Chiara Lupi and Pierangelo Geppetti

Abstract Ever since their identification, interest in the role of transient receptor potential (TRP) channels in health and disease has steadily increased. Robust evidence has underlined the role of TRP channels expressed in a subset of primary sensory neurons of the trigeminal ganglion to promote, by neuronal excitation, nociceptive responses, allodynia and hyperalgesia. In particular, the TRP vanilloid 1 (TRPV1) and the TRP ankyrin 1 (TRPA1) are expressed in nociceptive neurons, which also express the sensory neuropeptides, tachykinins, and calcitonin -related peptide (CGRP), which mediate neurogenic inflammatory responses. Of interest, CGRP released from the trigeminovascular network of neurons is currently recognized as a main contributing mechanism of migraine attack. The ability of TRPA1 to sense and to be activated by an unprecedented series of exogenous and endogenous reactive molecules has now been extensively documented. Several of the TRPA1 activators are also known as triggers of migraine attack. Thus, TRP channels, and particularly TRPA1, may be proposed as novel pathways in migraine pathophysiology and as possible new targets for its treatment. Keywords: Migraine; Transient receptor potential ankyrin 1 (TRPA1); Transient receptor potential vanilloid 1 (TRPV1); Calcitonin gene-related peptide (CGRP); Neurogenic inflammation; ThermoTRP; Headache; Pain

Review commonly described as nonselective Ca2+-permeable TRP channels channels, however, their Ca2+/Na+ permeability ratio The observation that the stimulation of a specific receptor, may vary widely between different members of the TRP and the consequent, associated transient inward current family [5]. [1,2] are necessary to vision in Drosophila melanogaster TRP channel gating is operated by both the direct has been the primal evidence of the transient receptor action on the channel of a plethora of exogenous and potential (TRP) family of channels, which currently endogenous physicochemical stimuli, and changes in encompasses more than 50 different channels [3]. TRP the intracellular machinery, including activation of channels represent a heterogeneous system oriented G- coupled receptor (GPCR) or tyrosine kinase towards environment perception, and participating in receptor [6]. In mammals, the TRP family consists of sensing visual, gustatory, olfactive, auditive, mechanical, 28 grouped into 6 subfamilies according to thermal, and osmotic stimuli. TRP channels consist of sequence identity, namely TRP canonical (TRPC), TRP six transmembrane domains (S1-S6) with both the NH2 vanilloid (TRPV), TRP melastatin (TRPM), TRP polycystin and COOH termini localized into the cytosol. The COOH (TRPP), TRP mucolipin (TRPML), and TRP ankyrin (TRPA) region is highly conserved among TRPs, whereas the [7,8]. The mammalian TRPC subfamily enlists 7 members NH2 region usually contains different numbers of (TRPC1-7), activated by the stimulation of GPCR and ankyrin repeats, the 33-residue motifs with a conserved receptor tyrosine kinases [9], although TRPC1 seems to be backbone and variable residues that mediate protein- directly activated by membrane stretch [10]. The TRPM protein interactions [4]. The entry of cations through has 8 members (TRPM1-8), and TRPM8 is activated by homo- or heterotetramers occurs via the pore formed and low temperatures (< 25°C). Of the three by loops between the S5 and S6 domains. TRPs are TRPML members, TRPML1 is widely expressed and has been described as an H+-sensor of endosomes/lyso- somes, where it probably prevents overacidification * Correspondence: [email protected] [11]. The TRPP family can be subdivided into PKD1- Headache Center and Clinical Pharmacology Unit, Department of Health Sciences, Careggi University Hospital, University of Florence, viale Pieraccini 6, like (TRPP1-like) and PKD2-like (TRPP2-like) proteins, Florence 50139, Italy

© 2013 Benemei et al.; licensee Springer. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Benemei et al. The Journal of Headache and Pain 2013, 14:71 Page 2 of 8 http://www.thejournalofheadacheandpain.com/content/14/1/71

which couple to act as a signaling complex at the plasma as a sensor of mechanical stimuli and noxious cold (< 17°C) membrane plasma membrane [12]. Various diseases have remains unresolved [36]. Electrophilic molecules have been attributed to mutations of TRP channels. However, been found to activate TRPA1 via auniquemechanism, only a few TRP channelopathies, commonly known as mediated by a Michael addition with specific cysteine “TRPpathies”, have been conclusively identified so far. and lysine residues identified in both the rat and TRPpathies include neurological disorders, renal diseases, channel [46,47]. Finally, TRPA1-expressing neurons also and complex skeletal dysplasias [13]. express other TRP channels, in particular TRPV1, and, even more importantly, the sensory neuropeptides substance TRPA1 and other thermo-TRPs P (SP), neurokinin A (NKA), and calcitonin gene-related Primary sensory neurons express different TRP channels, peptide (CGRP). SP/NKA and CGRP release from per- including four of the six members of the TRPV subfamily. ipheral endings of nociceptors promoted by TRPV1 or TRPV1, TRPV2, TRPV3, and TRPV4 channels sense TRPA1 activation produces a series of responses col- warm-hot temperatures and are chemosensors for a lectively described as neurogenic inflammation [48]. large series of naturally occurring and synthetic ligands. TRPV1, the receptor of the vanilloid compound TRPA1, pain and neurogenic inflammation (which promoted the labeling of the group with the V), It is generally recognized that C and Aδ-fibre sensory is responsive to high proton concentrations (pH 5–6) neurons convey pain signals, while neurons with larger- [14,15], [16] and various lipid derivatives size fibres mediate touch sensation. Thermo-TRPs, which [17]. and hypotonic solutions are non-selective under normal conditions are expressed in C and Aδ-fibre activators of the TRPV3 and TRPV4, respectively [18,19]. neurons, have been proposed to contribute to transmis- The synthetic compound 4α-phorbol 12,13-didecanoate sion and modulation of nociceptive signals. This conclu- (4α-PDD), low pH, citrate, endocannabinoids, and ara- sion originates from empirical observation that TRPV1 chidonic acid metabolites may also gate TRPV4 [20,21]. or TRPA1 agonists, derived from foods and spices, are Activators of TRPV2 are not well identified, although able to cause, in a dose-dependent fashion, a range from the uricosuric agent can activate the channel appreciated hot feelings to unpleasant pain sensation, [22]. Additional TRPs expressed in nociceptors are TRPA1 as in the case of capsaicin, the selective TRPV1 agonist and TRPM8. Finally, there is also evidence that TRPM3, contained in hot peppers, and , another TRPV1 rather uniquely activated by , seems agonist present in , or allyl , to be also expressed in primary sensory neurons [23]. the TRPA1 agonist contained in or [5,49]. A large NH2-terminal domain with 17 predicted ankyrin Another finding that enlists thermo-TRPs as major pain repeat domains characterizes TRPA1, the sole member controlling mechanisms is the clinical use of topical (cuta- of the TRPA subfamily. TRPA1, first cloned from human neous) capsaicin application that, by defunctionalizing fetal lung fibroblasts, is widely expressed in mammals, sensory nerve terminals, alleviates several pain conditions, where it has been found in hair cells, pancreas, heart, including post-herpetic neuralgias or pain associated with brain, keratinocytes [24], urinary bladder [25], prostate diabetic neuropathy [50]. [26], arteries [27], enterochromaffin cells [28], odonto- Generation of deleted mice and, more importantly, blasts and dental pulp [29,30], synovial fibroblasts [31], identification and preclinical development of selective and epithelial and smooth muscle cells of the airways antagonists for thermo-TRPs, have greatly increased our and lung [32]. A large amount of evidence shows that knowledge of the role of these channels in the regulation TRPA1 plays a key role in the detection of pungent or of acute nociceptive responses and the development of irritant compounds, including principles contained in allodynia and hyperalgesia. While TRPV1-deleted mice different spicy foods, such as (mustard exhibit decreased hyperalgesia to elevated temperatures oil) in [33], and diallyldisulfide in [51], TRPA1-deletion abrogated the two classical noci- [34], and in [35]. ceptive phases to formalin [38]. However, gene deletion (in ), (in ), (in may not completely recapitulate the effects of channel ), (in ), (in and blockade, as compensatory mechanism may counterbalance oregano) [36], are also able to gate TRPA1. In addition, the function of the absent gene-protein. Thus, findings environmental irritants and industry pollutants, such as produced by using selective thermo-TRP antagonists acetaldehyde, formalin, , hypochlorite, may better unveil the role of these channels in models isocyanates, ozone, carbon dioxide, ultraviolet light, and of pain diseases. (a highly reactive α,β-unsatured aldehyde present For detailed information on other thermo-TRPs, the in , cigarette smoke, smoke from burning vegeta- reader is referred to previous review articles [5-8] whereas tion, and vehicle exhaust), have been recognized as TRPA1 we will focus on the increasing evidence that supports activators [37-45]. The dispute regarding the role of TRPA1 the role of TRPA1 in models of both inflammatory and Benemei et al. The Journal of Headache and Pain 2013, 14:71 Page 3 of 8 http://www.thejournalofheadacheandpain.com/content/14/1/71

neuropathic pain. Mechanical hyperalgesia [52] and ongoing In addition, TRPA1 antagonists could represent novel neuronal discharge [53,54] evoked by complete Freund’s therapeutic strategies of CIPN. In this context, it could adjuvant (CFA) and tumour necrosis factor-α (TNFα), be better understood that the unexpected attenuation and mechanical hyperalgesia induced by low doses of by etodolac, a nonsteroidal anti-inflammatory drug of monosodium iodoacetate (MIA) [55] are inhibited by mechanical allodynia in a mouse model of neuropathic TRPA1 receptor antagonists in rodents. Convergent find- pain, might be due to its ability to inhibit TRPA1 [67]. ings indicate that TRPA1 contributes to carrageenan- In addition, to contribute to mechanical hyperalgesia, evoked inflammatory hyperalgesia [56,57]. Carrageenan TRPA1 seems to be involved in the development of cold administration, among a number of lipid derivatives, allodynia [68]. Cold allodynia was reduced in models of notably increases metabolites of 12-lipoxygenases, par- neuropathic (peripheral nerve injury) and inflammatory ticularly A3 (HXA3) and HXB3, whose hyper- (CFA) pain [69,70] or in models of CIPN, which typically algesic/allodynic effect is abrogated by TRPA1 antagonism exhibit this type of hypersensitivity [64-66]. Interestingly, [58]. It has also been found that TRPA1 mediates ongoing a familial episodic pain syndrome has been attributed nociception in chronic pancreatitis [59], and that both toagainoffunctionmutationofTRPA1TRPA1[71]. TRPV1 and TRPA1 initiate key pathways to transform Coincidence between TRPA1 and neuropeptide expres- acute into chronic inflammation and hyperalgesia in sion has been suggested [72,73], although evidence for pancreatitis [60]. The clinical observation that an anti- coexistence of isolectin B4-positive and non-peptidergic oxidant afforded protection in patients with pancreatitis neurons with TRPA1 has also been reported [74,75]. [61] indirectly supports the role of the oxidative stress Notwithstanding, exposure of tissues containing either sensor, TRPA1, in this condition. peripheral or central sensory nerve terminals to TRPA1 The underlying mechanisms that from neural tissue agonists invariably results in a -dependent release injury produce the chronic allodynia and hyperalgesia of SP/NKA and CGRP. Thus, TRPA1 stimulation has been typical of neuropathic pain are largely unknown. How- proven to increase sensory neuropeptide release from ever, recent reports have pointed to the role of TRPA1 oesophagus and urinary bladder, meninges, or dorsal in different models of neuropathic pain. Several meta- spinal cord [76-78]. The outcome of such a release in bolic pathways in glycolysis or lipid peroxidation pro- peripheral tissues encompasses the series of responses duce (MG), which appears in the plasma commonly referred to as ‘neurogenic inflammation’ [48] in diabetic patients as hyperglycemia strongly enhances (Figure 1). Although implication in transmission of noci- MG accumulation. MG has been recently described to ceptive signals has been proposed, the pathophysiological react reversibly with cysteine residues, and probably due outcome of the central release of sensory neuropeptides to this property stimulates TRPA1, thus representing a within the dorsal spinal cord or brain stem is less clear likely candidate metabolite to promote neuropathic pain (Figure 1). in metabolic disorders [62]. Chemotherapeutic induced peripheral neuropathy (CIPN) is a scarcely understood TRPA1, TRPV1, and migraine and poorly treated condition, which, characterized by One of the first findings that, although indirectly, sug- spontaneous pain, and mechanical and cold allodynia and gested the role of TRP channels in cluster headache and hyperalgesia, causes significant discomfort, and often migraine was represented by the protective effect of the therapy discontinuation. CIPN may outlast the time topical, desensitizing application of capsaicin to the pa- period of chemotherapeutic drug administration for weeks tient nasal mucosa [79,80]. Capsaicin treatment couples or months [63]. the unique ability of the drug to first activate, and, Alteration of several ion channels has been advocated subsequently, upon repeated administration, desensitize to explain this painful condition, but a univocal consensus both the afferent pathway that from channel activation has not emerged. Recently, in mouse models of CIPN conveys nociceptive signals, and the ‘efferent’ function produced by a single administration of oxaliplatin, pac- that results in sensory neuropeptide release [48]. Within litaxel or bortezomib, TRPA1 has been shown to play a the fifth cranial nerve, capsaicin desensitization causes major role in the development and maintenance of cold the defunctionalization of peripheral and possibly central and mechanical [64-66]. In a therapeutic perspective, it nerve endings, thus preventing SP/NKA-dependent plasma is of relevance that treatment with a TRPA1 antagonist protein extravasation within the dura mater, CGRP- just before and shortly after (about 6 hours) the admin- dependent dilatation of meningeal arterioles, and inhib- istration of bortezomib or oxaliplatin totally prevented ition of afferent nociceptive impulses. Further support the development and maintenance (for 10–15 days) of to the contribution of TRPV1 to migraine mechanism mechanical and cold hypersensitivity [66]. This finding derived from the observation that ethanol, a known suggests that TRPA1 is key in initiating CIPN and promot- trigger of migraine attacks, activates CGRP release from ing the transition from an acute to a chronic condition. sensory neurons, and promotes CGRP-dependent menin- Benemei et al. The Journal of Headache and Pain 2013, 14:71 Page 4 of 8 http://www.thejournalofheadacheandpain.com/content/14/1/71

Figure 1 Schematic representation of the probable mechanisms of the action of antimigraine remedies, either currently used, or proven effective in clinical trials (gray boxes) or of novel medicines (empty box), regarding their ability to modulate the release of calcitonin gene related peptide (CGRP) or the activation of its receptor (CGRP-R). (1) Non steroidal antiinflammatory drugs (NSAIDs) block prostaglandin synthesis and the ensuing nociceptor sensitization and CGRP release evoked by prostaglandin receptor (PG-R) activation. (2)

Triptans, by activating neuronal 5HT1D receptors, inhibit CGRP release. (3) CGRP-R antagonists inhibit the action of CGRP on effector cells. (4) Antagonists of transient receptor potential channels (TRPs), including TRP ankyrin 1 (TRPA1), block the ability of a series of stimulants (for TRPA1, cigarette smoke, acrolein, nitric oxide, , and others) to release CGRP. All medicines may act at both peripheral and central endings of trigeminal nociceptors. Receptor/channel activation may trigger/facilitate (+) or inhibit (−) CGRP release. geal vasodilatation by reducing the threshold temperature only 5–6 hours after nitroglycerine administration [92,93]. for channel activation, a phenomenon that eventually Although not confirmed in vitro [94]. NO may release results in TRPV1 activation [81,82]. CGRP from trigemino-vascular neurons [95]. More TRPV4 is stimulated by hypoosmotic stimuli that cause recently, NO has been revealed to target TRPA1 by plasma membrane stretch and mechanical distension. nitrosylation of channel cysteine residues [96], that Although this effect could, in principle, be implicated seem to differ from those targeted by other reactive in the throbbing pain often described by migraine patients molecules [97], and this novel molecular mechanism during their headache attacks, no evidence has yet been could contribute to the nociceptive response evoked by reported in support of TRPV4 in any model of head pain. NO [98]. It is possible that the S-nitrosylation process In contrast, a series of observational findings has recently [99], produced by NO, contributes to channel sensitization been obtained regarding a possible association between to eventually (hours after the exposure to NO) amplify TRPA1 and migraine. In this respect, it is of interest CGRP release by other agents, thus leading to exagger- that a number of compounds, recently identified as TRPA1 ated neurogenic inflammation and potentiation of pain agonists, including cigarette smoke, ammonium chloride, responses. , chlorine, garlic and others [34,38,41,78,83] The environmental pollution agent, acrolein, is pro- are known triggers of migraine attacks in susceptible duced by the combustion of organic material which causes individuals [84-89]. its accidental inhalation, which, however, occurs also with Nitric oxide (NO) donor drugs, including nitroglycerine, cigarette smoking [100]. Several components of cigarette are known inducers of migraine attacks and have been smoke, such as acrolein, crotonaldehyde [78], acetaldehyde extensively used in migraine provocation studies in [37] and [101], are TRPA1 agonists. Recently, [90]. Although their pro-migraine action has acrolein application to the rat nasal mucosa has been been attributed to their intrinsic vasodilatatory action shown to produce ipsilateral meningeal vasodilatation [91], the temporal mismatch between vasodilatation by a TRPA1- and CGRP-dependent mechanism [102], and the onset of migraine-like attacks argues against a thus offering a mechanistic explanation for the association close association between the two phenomena. In fact, between the exposure to cigarette smoke and migraine at the time of the maximum vasodilatation, a mild to attack appearance or worsening [103,104]. moderate headache develops both in migraineurs (more) Herbalism has been instrumental for the development and in healthy subjects (less), while delayed migraine-like of pharmacology and also to a better understanding of attacks, present only in migraine patients, are observed pathophysiological mechanisms. These principles also Benemei et al. The Journal of Headache and Pain 2013, 14:71 Page 5 of 8 http://www.thejournalofheadacheandpain.com/content/14/1/71

apply to the migraine field. Umbellularia californica graine, as compared to pain‐free state, and that topical (California bay laurel) is also known as the ‘headache application of inflammatory molecules on the rat menin- tree’ because of the ability of its scent to trigger headache ges sensitizes thalamic trigeminovascular neurons [112]. attacks in susceptible individuals [105]. A case of cluster Each component of the nociceptive pathways could con- headache-like attacks preceded by cold sensations per- tribute differently to sensitization of the neural tissues. ceived in the ipsilateral nostril following inhalation of TRP channels could be sensitized by different compounds Umbellularia californica scent has recently been described and via different mechanisms [5,36,66,81,113]. Although in a cluster headache patient whose attacks had ceased there is no specific information on the role of these chan- 10 years before [106]. The irritant ketone, nels to peripheral nociceptor sensitization or central sensi- umbellulone, one of the most abundant reactive mole- tization in migraine, it is possible that TRP channels, and cules of Umbellularia californica, was found to activate in particular TRPV1 and TRPA1, contribute to this key the human recombinant and the constitutive rat/mouse mechanism. TRPA1 in trigeminal ganglia (TG) neurons, and via this mechanism to produce nociceptive behaviour and the Conclusions release of CGRP from TG or meningeal tissue in rats [77]. The still largely unfinished puzzle of the migraine mech- In addition, similar to acrolein, umbellulone application anism is being completed by novel unexpected pieces of to the rat nasal mucosa evoked ipsilateral TRPA1- and information, which compose a clearer picture. The first, CGRP-dependent meningeal vasodilatation [77]. corner of the picture, known for decades, is that cyclo- Ligustilide, an electrophilic volatile dihydrophthalide of oxygenase (namely cyclooxygenase 2) inhibition has a dietary and medicinal relevance, has been found to beneficial effect on migraine attack. The second corner, produce a moderate activation of TRPA1, but also to predicted by scientists and appreciated by clinicians and inhibit allyl isothiocyanate-evoked stimulation of TRPA1 patients, is that targeting 5-HT1 receptors is also highly [107]. This newly identified target of ligustilide offers a effective. The third corner is that a series of clinical trials novel mechanistic explanation for the use of the compound show that CGRP antagonists afford a protection similar in traditional medicine to treat pain diseases, including to that of triptans. To complete at least the frame, a headaches. Finally, although several hypotheses have fourth corner, which should reconcile the other three in been advanced, the mechanism of the analgesic action of an intelligible sequence of events, is needed. While pros- acetaminophen () in different pain conditions taglandins sensitize nociceptors and eventually promote is far from clear. The reactive metabolite of acetamino- CGRP release, triptans inhibit such release, thus produ- phen, N-acetyl-p-benzo-quinoneimine (NAPQI), is able to cing an indirect anti-migraine effect, and CGRP antago- activate the TRPA1 channel and thereby evoke a moderate nists abrogate the final common pathway of migraine and reversible neurogenic inflammatory response, which, mechanism. The pathway, which, sensitized by prosta- in susceptible individuals, may contribute to emphasizing glandins and inhibited by serotonin receptor stimulation, inflammation in peripheral tissues [76]. However, NAPQI results in trigeminal neuron activation and the pro- may also be produced by cytochrome activity within migraine release of CGRP could represent the fourth the spinal cord, and NAPQI action at the spinal level corner of the picture. Emerging information on TRP chan- results in channel desensitization [108]. Inhibition of nels, and particularly TRPA1, which, targeted by migraine central TRPA1 has been advocated as the mechanism, triggers, contribute, by activating the trigeminal CGRP- which may be responsible the hitherto unexplained an- dependent pathway, to the genesis of pain and the accom- algesic and possibly antimigraine action of its parent panying symptoms of the attack, seems to be of paramount molecule [108]. Importantly, this novel spinal mechanism importance to solve what still remains the enigma of the could be of general relevance also for other TRPA1 migraine mechanism. agonists which share with NAPQI the ability of de- sensitizing the channel. Competing interests A host of endogenous inflammatory mediators possibly The authors declare that they have no competing interests. released during migraine attacks can activate and sensitize peripheral and central sensory neurons, including tri- Authors’ contribution geminal neurons. Sensitization of first-order neurons is FDC, CF, ER, and CL searched for the literature. SB and PG wrote and revised involved in the perception of headache throbbing pain the text. All authors read and approved the final manuscript. [109], while sensitization of second‐order neurons con- tributes to cephalic allodynia and muscle tenderness Acknowledgements [110,111]. Recently, it has been shown that innocuous We acknowledge Regione Toscana (Regional Health Research Program 2009, P.G.). brush and heat stimuli induce larger activation in the Received: 22 May 2013 Accepted: 10 August 2013 thalamus of patients who exhibit allodynia during mi- Published: 13 August 2013 Benemei et al. The Journal of Headache and Pain 2013, 14:71 Page 6 of 8 http://www.thejournalofheadacheandpain.com/content/14/1/71

References receptor activity in human normal and hyperplastic prostate: relation to 1. Minke B (1977) Drosophila mutant with a transducer defect. Biophys Struct nerves and interstitial cells. Eur Urol 57:902–910 Mech 3:59–64 27. Earley S, Gonzales AL, Crnich R (2009) Endothelium-dependent cerebral 2. Montell C, Jones K, Hafen E, Rubin G (1985) Rescue of the Drosophila artery dilation mediated by TRPA1 and Ca2+−Activated K+ channels. phototransduction mutation trp by germline transformation. Science Circ Res 104:987–994 230:1040–1043 28. Nozawa K, Kawabata-Shoda E, Doihara H, Kojima R, Okada H, Mochizuki S, 3. Vriens J, Owsianik G, Voets T, Droogmans G, Nilius B (2004) Invertebrate TRP Sano Y, Inamura K, Matsushime H, Koizumi T, Yokoyama T, Ito H (2009) proteins as functional models for mammalian channels. Pflugers Arch TRPA1 regulates gastrointestinal motility through serotonin release from 449:213–226 enterochromaffin cells. Proc Natl Acad Sci USA 106:3408–3413 4. Sedgwick SG, Smerdon SJ (1999) The ankyrin repeat: a diversity of interactions 29. El Karim IA, Linden GJ, Curtis TM, About I, McGahon MK, Irwin CR, Lundy FT on a common structural framework. Trends Biochem Sci 24:311–316 (2010) Human odontoblasts express functional thermo-sensitive TRP 5. Nilius B, Owsianik G, Voets T, Peters JA (2007) Transient receptor potential channels: implications for dentin sensitivity. Pain 152:2211–2223 cation channels in disease. Physiol Rev 87:165–217 30. El Karim IA, Linden GJ, Curtis TM, About I, McGahon MK, Irwin CR, 6. Ramsey IS, Delling M, Clapham DE (2006) An introduction to TRP channels. Killough SA, Lundy FT (2011) Human dental pulp fibroblasts express the Annu Rev Physiol 68:619–647 “cold-sensing” transient receptor potential channels TRPA1 and TRPM8. 7. Clapham DE (2003) TRP channels as cellular sensors. Nature 426:517–524 J Endod 37:473–478 8. Montell C, Birnbaumer L, Flockerzi V (2002) The TRP channels, a remarkably 31. Kochukov MY, McNearney TA, Fu Y, Westlund KN (2006) Thermosensitive functional family. Cell 108:595–598 TRP ion channels mediate cytosolic calcium response in human 9. Montell C (1999) Visual transduction in Drosophila. Annu Rev Cell Dev Biol synoviocytes. Am J Physiol Cell Physiol 291:C424–C432 15:231–268 32. Nassini R, Pedretti P, Moretto N, Fusi C, Carnini C, Facchinetti F, Viscomi 10. Maroto R, Raso A, Wood TG, Kurosky A, Martinac B, Hamill OP (2005) TRPC1 AR, Pisano AR, Stokesberry S, Brunmark C, Svitacheva N, McGarvey L, forms the stretch-activated cation channel in vertebrate cells. Nat Cell Biol Patacchini R, Damholt AB, Geppetti P, Materazzi S (2012) Transient 7:179–185 receptor potential ankyrin 1 channel localized to non-neuronal airway 11. Soyombo AA, Tjon-Kon-Sang S, Rbaibi Y, Bashllari E, Bisceglia J, Muallem S, cells promotes non-neurogenic inflammation. PLoS One 7:e42454 Kiselyov K (2006) TRP-ML1 regulates lysosomal pH and acidic lysosomal lipid 33. Jordt SE, Bautista DM, Chuang HH, McKemy DD, Zygmunt PM, Hogestatt hydrolytic activity. J Biol Chem 281:7294–7301 ED, Meng ID, Julius D (2004) Mustard oils and excite sensory 12. Hanaoka K, Qian F, Boletta A, Bhunia AK, Piontek K, Tsiokas L, Sukhatme VP, nerve fibres through the TRP channel ANKTM1. Nature 427:260–265 Guggino WB, Germino GG (2000) Co-assembly of polycystin-1 and −2 34. Bautista DM, Movahed P, Hinman A, Axelsson HE, Sterner O, Hogestatt ED, produces unique cation-permeable currents. Nature 408:990–994 Julius D, Jordt SE, Zygmunt PM (2005) Pungent products from garlic 13. Nilius B, Owsianik G (2010) Transient receptor potential channelopathies. activate the sensory TRPA1. Proc Natl Acad Sci USA Pflugers Arch 460:437–450 102:12248–12252 14. Bevan S, Geppetti P (1994) Protons: small stimulants of capsaicin-sensitive 35. Bandell M, Story GM, Hwang SW, Viswanath V, Eid SR, Petrus MJ, Earley TJ, sensory nerves. Trends Neurosci 17:509–512 Patapoutian A (2004) Noxious cold ion channel TRPA1 is activated by 15. Tominaga M, Caterina MJ, Malmberg AB, Rosen TA, Gilbert H, Skinner K, pungent compounds and . Neuron 41:849–857 Raumann BE, Basbaum AI, Julius D (1998) The cloned capsaicin receptor 36. Nilius B, Appendino G, Owsianik G (2012) The transient receptor potential integrates multiple pain-producing stimuli. Neuron 21:531–543 channel TRPA1: from gene to pathophysiology. Pflugers Arch 464:425–458 16. Zygmunt PM, Petersson J, Andersson DA, Chuang H, Sorgard M, Di Marzo V, 37. Bang S, Kim KY, Yoo S, Kim YG, Hwang SW (2007) Transient receptor Julius D, Hogestatt ED (1999) Vanilloid receptors on sensory nerves mediate potential A1 mediates acetaldehyde-evoked pain sensation. Eur J Neurosci the vasodilator action of anandamide. Nature 400:452–457 26:2516–2523 17. Shin J, Cho H, Hwang SW, Jung J, Shin CY, Lee SY, Kim SH, Lee MG, Choi 38. McNamara CR, Mandel-Brehm J, Bautista DM, Siemens J, Deranian KL, Zhao YH, Kim J, Haber NA, Reichling DB, Khasar S, Levine JD, Oh U (2002) M, Hayward NJ, Chong JA, Julius D, Moran MM, Fanger CM (2007) TRPA1 Bradykinin-12-lipoxygenase-VR1 signaling pathway for inflammatory mediates formalin-induced pain. Proc Natl Acad Sci USA 104:13525–13530 hyperalgesia. Proc Natl Acad Sci USA 99:10150–10155 39. Andersson DA, Gentry C, Moss S, Bevan S (2008) Transient receptor 18. Moqrich A, Hwang SW, Earley TJ, Petrus MJ, Murray AN, Spencer KS, Andahazy potential A1 is a sensory receptor for multiple products of oxidative stress. M, Story GM, Patapoutian A (2005) Impaired thermosensation in mice lacking J Neurosci 28:2485–2494 TRPV3, a heat and camphor sensor in the skin. Science 307:1468–1472 40. Sawada Y, Hosokawa H, Matsumura K, Kobayashi S (2008) Activation of 19. Everaerts W, Nilius B, Owsianik G (2010) The vanilloid transient receptor potential transient receptor potential ankyrin 1 by hydrogen peroxide. Eur J Neurosci channel TRPV4: from structure to disease. Prog Biophys Mol Biol 103:2–17 27:1131–1142 20. Watanabe H, Vriens J, Prenen J, Droogmans G, Voets T, Nilius B (2003) 41. Bessac BF, Sivula M, von Hehn CA, Escalera J, Cohn L, Jordt SE (2008) TRPA1 Anandamide and use epoxyeicosatrienoic acids to activate is a major oxidant sensor in murine airway sensory neurons. J Clin Invest TRPV4 channels. Nature 424:434–438 118:1899–1910 21. Vriens J, Owsianik G, Fisslthaler B, Suzuki M, Janssens A, Voets T, Morisseau 42. Bessac BF, Sivula M, von Hehn CA, Caceres AI, Escalera J, Jordt SE (2009) C, Hammock BD, Fleming I, Busse R, Nilius B (2005) Modulation of the Ca2 Transient receptor potential ankyrin 1 antagonists block the noxious effects permeable cation channel TRPV4 by cytochrome P450 epoxygenases in of toxic industrial isocyanates and tear gases. Faseb J 23:1102–1114 vascular endothelium. Circ Res 97:908–915 43. Taylor-Clark TE, Undem BJ (2010) Ozone activates airway nerves via the 22. Bang S, Kim KY, Yoo S, Lee SH, Hwang SW (2007) Transient receptor selective stimulation of TRPA1 ion channels. J Physiol 588:423–433 potential V2 expressed in sensory neurons is activated by probenecid. 44. Wang YY, Chang RB, Liman ER (2010) TRPA1 is a component of the Neurosci Lett 425:120–125 nociceptive response to CO2. J Neurosci 30:12958–12963 23. Wagner TF, Loch S, Lambert S, Straub I, Mannebach S, Mathar I, Dufer M, Lis 45. Hill K, Schaefer M (2009) Ultraviolet light and photosensitising agents A, Flockerzi V, Philipp SE, Oberwinkler J (2008) Transient receptor potential activate TRPA1 via generation of oxidative stress. Cell Calcium 45:155–164 M3 channels are ionotropic receptors in pancreatic beta cells. 46. Macpherson LJ, Dubin AE, Evans MJ, Marr F, Schultz PG, Cravatt BF, Nat Cell Biol 10:1421–1430 Patapoutian A (2007) Noxious compounds activate TRPA1 ion channels 24. Atoyan R, Shander D, Botchkareva NV (2009) Non-neuronal expression of through covalent modification of cysteines. Nature 445:541–545 transient receptor potential type A1 (TRPA1) in human skin. J Invest 47. Hinman A, Chuang HH, Bautista DM, Julius D (2006) TRP channel activation by Dermatol 129:2312–2315 reversible covalent modification. Proc Natl Acad Sci USA 103:19564–19568 25. Streng T, Axelsson HE, Hedlund P, Andersson DA, Jordt SE, Bevan S, 48. Geppetti P, Holzer P (1996) Neurogenic inflammation. CRC Press, Boca Raton Andersson KE, Hogestatt ED, Zygmunt PM (2008) Distribution and function 49. Preti D, Szallasi A, Patacchini R (2012) TRP channels as therapeutic targets in of the hydrogen sulfide-sensitive TRPA1 ion channel in rat urinary bladder. airway disorders: a patent review. Expert Opin Ther Pat 22:663–695 Eur Urol 53:391–399 50. Derry S, Sven-Rice A, Cole P, Tan T, Moore RA (2013) Topical capsaicin 26. Gratzke C, Weinhold P, Reich O, Seitz M, Schlenker B, Stief CG, Andersson (high concentration) for chronic neuropathic pain in adults. Cochrane KE, Hedlund P (2010) Transient receptor potential A1 and Database Syst Rev 2, CD007393 Benemei et al. The Journal of Headache and Pain 2013, 14:71 Page 7 of 8 http://www.thejournalofheadacheandpain.com/content/14/1/71

51. Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, Julius D McDonald HA, Schmidt RG, McGaraughty SP, Chu KL, Faltynek CR, Kort ME, (1997) The capsaicin receptor: a heat-activated ion channel in the pain Reilly RM, Kym PR (2011) Selective blockade of TRPA1 channel attenuates pathway. Nature 389:816–824 pathological pain without altering noxious cold sensation or body 52. Fernandes ES, Russell FA, Spina D, McDougall JJ, Graepel R, Gentry C, temperature regulation. Pain 152:1165–1172 Staniland AA, Mountford DM, Keeble JE, Malcangio M, Bevan S, Brain SD 71. Kremeyer B, Lopera F, Cox JJ, Momin A, Rugiero F, Marsh S, Woods CG, (2011) A distinct role for transient receptor potential ankyrin 1, in addition Jones NG, Paterson KJ, Fricker FR, Villegas A, Acosta N, Pineda-Trujillo NG, to transient receptor potential vanilloid 1, in tumor necrosis factor Ramirez JD, Zea J, Burley MW, Bedoya G, Bennett DL, Wood JN, Ruiz-Linares alpha-induced inflammatory hyperalgesia and Freund’s complete A (2010) A gain-of-function mutation in TRPA1 causes familial episodic pain adjuvant-induced monarthritis. Arthritis Rheum 63:819–829 syndrome. Neuron 66:671–680 53. McGaraughty S, Chu KL, Perner RJ, Didomenico S, Kort ME, Kym PR (2010) 72. Bhattacharya MR, Bautista DM, Wu K, Haeberle H, Lumpkin EA, Julius D (2008) TRPA1 modulation of spontaneous and mechanically evoked firing of spinal Radial stretch reveals distinct populations of mechanosensitive mammalian neurons in uninjured, osteoarthritic, and inflamed rats. Mol Pain 6:14 somatosensory neurons. Proc Natl Acad Sci USA 105:20015–20020 54. da Costa DS, Meotti FC, Andrade EL, Leal PC, Motta EM, Calixto JB (2010) 73. Story GM, Peier AM, Reeve AJ, Eid SR, Mosbacher J, Hricik TR, Earley TJ, The involvement of the transient receptor potential A1 (TRPA1) in the Hergarden AC, Andersson DA, Hwang SW, McIntyre P, Jegla T, Bevan S, maintenance of mechanical and cold hyperalgesia in persistent Patapoutian A (2003) ANKTM1, a TRP-like channel expressed in nociceptive inflammation. Pain 148:431–437 neurons, is activated by cold temperatures. Cell 112:819–829 55. Okun A, Liu P, Davis P, Ren J, Remeniuk B, Brion T, Ossipov MH, Xie J, 74. Barabas ME, Kossyreva EA, Stucky CL (2012) TRPA1 is functionally expressed Dussor GO, King T, Porreca F (2012) Afferent drive elicits ongoing pain in a primarily by IB4-binding, non-peptidergic mouse and rat sensory neurons. model of advanced osteoarthritis. Pain 153:924–933 PLoS One 7:e47988 56. Bonet IJ, Fischer L, Parada CA, Tambeli CH (2013) The role of transient 75. Kim YS, Son JY, Kim TH, Paik SK, Dai Y, Noguchi K, Ahn DK, Bae YC (2010) receptor potential A 1 (TRPA1) in the development and maintenance of Expression of transient receptor potential ankyrin 1 (TRPA1) in the rat trigeminal carrageenan-induced hyperalgesia. Neuropharmacology 65:206–212 sensory afferents and spinal dorsal horn. J Comp Neurol 518:687–698 57. Moilanen LJ, Laavola M, Kukkonen M, Korhonen R, Leppanen T, Hogestatt 76. Nassini R, Materazzi S, Andre E, Sartiani L, Aldini G, Trevisani M, Carnini C, ED, Zygmunt PM, Nieminen RM, Moilanen E (2012) TRPA1 contributes to Massi D, Pedretti P, Carini M, Cerbai E, Preti D, Villetti G, Civelli M, Trevisan G, the acute inflammatory response and mediates carrageenan-induced paw Azzari C, Stokesberry S, Sadofsky L, McGarvey L, Patacchini R, Geppetti P edema in the mouse. Sci Rep 2:380 (2010) Acetaminophen, via its reactive metabolite N-acetyl-p-benzo- 58. GregusAM,DoolenS,DumlaoDS,BuczynskiMW,TakasusukiT,FitzsimmonsBL, quinoneimine and transient receptor potential ankyrin-1 stimulation, causes Hua XY, Taylor BK, Dennis EA, Yaksh TL (2013) Spinal 12-lipoxygenase-derived neurogenic inflammation in the airways and other tissues in rodents. Faseb A3 contributes to inflammatory hyperalgesia via activation of TRPV1 J 24:4904–4916 and TRPA1 receptors. Proc Natl Acad Sci USA 109:6721–6726 77. Nassini R, Materazzi S, Vriens J, Prenen J, Benemei S, De Siena G, la Marca G, 59. CattaruzzaF,JohnsonC,LeggitA,GradyEF,SchenkAK,CevikbasF,CedronWJ, Andre E, Preti D, Avonto C, Sadofsky L, Di Marzo V, De Petrocellis L, Dussor Bondada S, Kirkwood R, Malone BJ, Steinhoff M, Bunnett NW, Kirkwood KS (2013) G, Porreca F, Taglialatela-Scafati O, Appendino G, Nilius B, Geppetti P (2012) Transient Receptor Potential Ankyrin 1 (TRPA1) Mediates Chronic Pancreatitis Pain The ‘headache tree’ via umbellulone and TRPA1 activates the In Mice. Am J Physiol Gastrointest Liver Physiol 304:G1002–G1012 trigeminovascular system. Brain 135:376–390 60. Schwartz ES, La JH, Scheff NN, Davis BM, Albers KM, Gebhart GF (2013) 78. Andre E, Campi B, Materazzi S, Trevisani M, Amadesi S, Massi D, Creminon C, TRPV1 and TRPA1 antagonists prevent the transition of acute to chronic Vaksman N, Nassini R, Civelli M, Baraldi PG, Poole DP, Bunnett NW, Geppetti inflammation and pain in chronic pancreatitis. J Neurosci 33:5603–5611 P, Patacchini R (2008) Cigarette smoke-induced neurogenic inflammation is 61. Bhardwaj P, Garg PK, Maulik SK, Saraya A, Tandon RK, Acharya SK (2009) A mediated by alpha, beta-unsaturated aldehydes and the TRPA1 receptor in randomized controlled trial of antioxidant supplementation for pain relief in rodents. J Clin Invest 118:2574–2582 patients with chronic pancreatitis. Gastroenterology 136:149–159 79. Fusco BM, Barzoi G, Agro F (2003) Repeated intranasal capsaicin applications 62. Eberhardt MJ, Filipovic MR, Leffler A, de la Roche J, Kistner K, Fischer MJ, to treat chronic migraine. Br J Anaesth 90:812 Fleming T, Zimmermann K, Ivanovic-Burmazovic I, Nawroth PP, Bierhaus A, 80. Fusco BM, Marabini S, Maggi CA, Fiore G, Geppetti P (1994) Preventative effect Reeh PW, Sauer SK (2012) Methylglyoxal activates nociceptors through of repeated nasal applications of capsaicin in cluster headache. Pain 59:321–325 transient receptor potential channel A1 (TRPA1): a possible mechanism of 81. Trevisani M, Geppetti P, Davis JB, Bianchi A, Harrison S, Randall AD, Smith metabolic neuropathies. J Biol Chem 287:28291–28306 GD, Owen D, Brough SJ, Jerman JC, Gray J, Amadesi S, Campi B, Barbieri M, 63. Cavaletti G, Marmiroli P (2010) Chemotherapy-induced peripheral Tognetto M, Gunthorpe MJ, Smart D (2002) Ethanol elicits and potentiates neurotoxicity. Nat Rev Neurol 6:657–666 nociceptor responses via the vanilloid receptor-1. Nat Neurosci 5:546–551 64. Nassini R, Gees M, Harrison S, De Siena G, Materazzi S, Moretto N, Failli P, 82. Nicoletti P, Trevisani M, Manconi M, Gatti R, De Siena G, Zagli G, Benemei S, Preti D, Marchetti N, Cavazzini A, Mancini F, Pedretti P, Nilius B, Patacchini R, Capone JA, Geppetti P, Pini LA (2008) Ethanol causes neurogenic Geppetti P (2011) Oxaliplatin elicits mechanical and cold allodynia in by TRPV1 activation and CGRP release in the trigeminovascular rodents via TRPA1 receptor stimulation. Pain 152:1621–1631 system of the guinea pig. Cephalalgia 28:9–17 65. Materazzi S, Fusi C, Benemei S, Pedretti P, Patacchini R, Nilius B, Prenen J, 83. Fujita F, Uchida K, Moriyama T, Shima A, Shibasaki K, Inada H, Sokabe T, Creminon C, Geppetti P, Nassini R (2012) TRPA1 and TRPV4 mediate Tominaga M (2008) Intracellular alkalization causes pain sensation through -induced peripheral neuropathy in mice via a glutathione-sensitive activation of TRPA1 in mice. J Clin Invest 118:4049–4057 mechanism. Pflugers Arch 463:561–569 84. Courteau JP, Cushman R, Bouchard F, Quevillon M, Chartrand A, Bherer L 66. Trevisan G, Materazzi S, Fusi C, Altomare A, Aldini G, Lodovici M, Patacchini (1994) Survey of construction workers repeatedly exposed to chlorine over R, Geppetti P, Nassini R (2013) Novel Therapeutic Strategy to Prevent a three to six month period in a pulpmill: I. Exposure and symptomatology. Chemotherapy-Induced Persistent Sensory Neuropathy By TRPA1 Blockade. Occup Environ Med 51:219–224 Cancer Res 73:3120–3131 85. Peatfield RC (1995) Relationships between food, wine, and beer-precipitated 67. Inoue N, Ito S, Nogawa M, Tajima K, Kyoi T (2012) Etodolac blocks the allyl migrainous headaches. Headache 35:355–357 isothiocyanate-induced response in mouse sensory neurons by selective 86. Wantke F, Focke M, Hemmer W, Bracun R, Wolf-Abdolvahab S, Gotz M, TRPA1 activation. Pharmacology 90:47–54 Jarisch R, Gotz M, Tschabitscher M, Gann M, Tappler P (2000) Exposure to 68. Karashima Y, Talavera K, Everaerts W, Janssens A, Kwan KY, Vennekens R, formaldehyde and phenol during an anatomy dissecting course: sensitizing Nilius B, Voets T (2009) TRPA1 acts as a cold sensor in vitro and in vivo. Proc potency of formaldehyde in medical students. Allergy 55:84–87 Natl Acad Sci USA 106:1273–1278 87. Irlbacher K, Meyer BU (2002) Nasally triggered headache. Neurology 58:294 69. del Camino D, Murphy S, Heiry M, Barrett LB, Earley TJ, Cook CA, Petrus MJ, 88. Kelman L (2007) The triggers or precipitants of the acute migraine attack. Zhao M, D’Amours M, Deering N, Brenner GJ, Costigan M, Hayward NJ, Cephalalgia 27:394–402 Chong JA, Fanger CM, Woolf CJ, Patapoutian A, Moran MM (2010) TRPA1 89. Roussos AP, Hirsch AR (2013) Alliaceous Migraines. Headache 2013:12091 contributes to cold hypersensitivity. J Neurosci 30:15165–15174 90. Iversen HK, Olesen J (1996) Headache induced by a nitric oxide donor 70. Chen J, Joshi SK, DiDomenico S, Perner RJ, Mikusa JP, Gauvin DM, Segreti (nitroglycerin) responds to sumatriptan. A human model for development JA, Han P, Zhang XF, Niforatos W, Bianchi BR, Baker SJ, Zhong C, Simler GH, of migraine drugs. Cephalalgia 16:412–418 Benemei et al. The Journal of Headache and Pain 2013, 14:71 Page 8 of 8 http://www.thejournalofheadacheandpain.com/content/14/1/71

91. Shevel E (2011) The extracranial vascular theory of migraine–a great story confirmed by the facts. Headache 51:409–417 92. Iversen HK, Olesen J (1994) Nitroglycerin-induced headache is not dependent on histamine release: support for a direct nociceptive action of nitric oxide. Cephalalgia 14:437–442 93. Thomsen LL, Olesen J (2001) Nitric oxide in primary headaches. Curr Opin Neurol 14:315–321 94. Eltorp CT, Jansen-Olesen I, Hansen AJ (2000) Release of calcitonin gene-related peptide (CGRP) from guinea pig dura mater in vitro is inhibited by sumatriptan but unaffected by nitric oxide. Cephalalgia 20:838–844 95. Wei EP, Moskowitz MA, Boccalini P, Kontos HA (1992) Calcitonin gene-related peptide mediates nitroglycerin and sodium nitroprusside-induced vasodilation in feline cerebral arterioles. Circ Res 70:1313–1319 96. Yoshida T, Inoue R, Morii T, Takahashi N, Yamamoto S, Hara Y, Tominaga M, Shimizu S, Sato Y, Mori Y (2006) Nitric oxide activates TRP channels by cysteine S-nitrosylation. Nat Chem Biol 2:596–607 97. Takahashi N, Mizuno Y, Kozai D, Yamamoto S, Kiyonaka S, Shibata T, Uchida K, Mori Y (2008) Molecular characterization of TRPA1 channel activation by cysteine-reactive inflammatory mediators. Channels (Austin) 2:287–298 98. Miyamoto T, Dubin AE, Petrus MJ, Patapoutian A (2009) TRPV1 and TRPA1 mediate peripheral nitric oxide-induced nociception in mice. PLoS One 4:e7596 99. Takahashi N, Mori Y (2011) TRP Channels as Sensors and Signal Integrators of Redox Status Changes. Front Pharmacol 2:58 100. Facchinetti F, Amadei F, Geppetti P, Tarantini F, Di Serio C, Dragotto A, Gigli PM, Catinella S, Civelli M, Patacchini R (2007) Alpha, beta-unsaturated aldehydes in cigarette smoke release inflammatory mediators from human macrophages. Am J Respir Cell Mol Biol 37:617–623 101. Talavera K, Gees M, Karashima Y, Meseguer VM, Vanoirbeek JA, Damann N, Everaerts W, Benoit M, Janssens A, Vennekens R, Viana F, Nemery B, Nilius B, Voets T (2009) Nicotine activates the chemosensory cation channel TRPA1. Nat Neurosci 12:1293–1299 102. Kunkler PE, Ballard CJ, Oxford GS, Hurley JH (2011) TRPA1 receptors mediate environmental irritant-induced meningeal vasodilatation. Pain 152:38–44 103. Lima AM, Sapienza GB, Giraud Vde O, Fragoso YD (2011) Odors as triggering and worsening factors for migraine in men. Arq Neuropsiquiatr 69:324–327 104. Friedman DI, De ver Dye T (2009) Migraine and the environment. Headache 49:941–952 105. California Laurel (2006) USDA Natural Resources Conservation Service., http://plants.usda.gov/plantguide/pdf/cs_umca.pdf. Accessed 16 Apr 2009 106. Benemei S, Appendino G, Geppetti P (2009) Pleasant natural scent with unpleasant effects: cluster headache-like attacks triggered by Umbellularia californica. Cephalalgia 30:744–746 107. Zhong J, Minassi A, Prenen J, Taglialatela-Scafati O, Appendino G, Nilius B (2011) Umbellulone modulates TRP channels. Pflugers Arch 462:861–870 108. Andersson DA, Gentry C, Alenmyr L, Killander D, Lewis SE, Andersson A, Bucher B, Galzi JL, Sterner O, Bevan S, Hogestatt ED, Zygmunt PM (2011) TRPA1 mediates spinal antinociception induced by acetaminophen and the cannabinoid Delta(9)-. Nat Commun 2:551 109. Strassman AM, Raymond SA, Burstein R (1996) Sensitization of meningeal sensory neurons and the origin of headaches. Nature 384:560–564 110. Burstein R, Cutrer MF, Yarnitsky D (2000) The development of cutaneous allodynia during a migraine attack clinical evidence for the sequential recruitment of spinal and supraspinal nociceptive neurons in migraine. Brain 123(Pt 8):1703–1709 111. Burstein R, Jakubowski M, Garcia-Nicas E, Kainz V, Bajwa Z, Hargreaves R, Becerra L, Borsook D (2010) Thalamic sensitization transforms localized pain into widespread allodynia. Ann Neurol 68:81–91 112. Noseda R, Burstein R (2013) Migraine pathophysiology: anatomy of the trigeminovascular pathway and associated neurological symptoms, CSD, Submit your manuscript to a sensitization and modulation of pain. Pain, Epub ahead of print journal and benefi t from: 113. Selescu T, Ciobanu AC, Dobre C, Reid G, Babes A (2013) Camphor activates and sensitizes transient receptor potential melastatin 8 (TRPM8) to cooling 7 Convenient online submission and . Chem Senses, Epub ahead of print 7 Rigorous peer review 7 Immediate publication on acceptance doi:10.1186/1129-2377-14-71 Cite this article as: Benemei et al.: TRPA1 and other TRP channels in 7 Open access: articles freely available online migraine. The Journal of Headache and Pain 2013 14:71. 7 High visibility within the fi eld 7 Retaining the copyright to your article

Submit your next manuscript at 7 springeropen.com